Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters










Database
Language
Publication year range
1.
Mil Med ; 184(Suppl 1): 282-290, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30901474

ABSTRACT

OBJECTIVE: Blast injury is associated with multi-organ failure (MOF), causing significant morbidity and mortality in trauma patients. However, the pathogenesis of blast-induced MOF still remains obscure. In this study, we evaluate the pathophysiological changes related to blast-induced MOF in a clinically relevant rat model of blast injury. METHODS: A moderate blast overpressure was applied to induce injury in anesthetized rats. Pathological changes were evaluated by H&E staining. Complement activation, plasminogen, and myeloperoxidase levels were analyzed by complement hemolytic assay (CH50) and/or ELISA in blood samples. RESULTS: Analysis of lung, brain, and liver tissue at 24 hour after blast overpressure revealed severe injuries. The level of complement components C3 and C1q decreased in parallel with the reduction of CH50 level in injured animals at 1, 3, and 6 hours after blast. Consumption of plasminogen was also detected as early as 1 hour post-injury. Myeloperoxidase levels were elevated within 1 hour of blast injury. CONCLUSION: Our data reveal that blast injury triggers the complement and fibrinolytic systems, which likely contribute to blast-induced MOF. Conceivably, therapies that target these systems early may improve clinical outcomes in blast patients.


Subject(s)
Blast Injuries/drug therapy , Fibrinolytic Agents/pharmacokinetics , Multiple Organ Failure/etiology , Animals , Blast Injuries/complications , Brain/drug effects , Brain/pathology , Brain/physiopathology , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay/methods , Fibrinolytic Agents/blood , Fibrinolytic Agents/therapeutic use , Injury Severity Score , Liver/drug effects , Liver/pathology , Liver/physiopathology , Lung/drug effects , Lung/pathology , Lung/physiopathology , Male , Multiple Organ Failure/drug therapy , Rats , Rats, Sprague-Dawley/blood , Statistics, Nonparametric
2.
PLoS One ; 13(8): e0202594, 2018.
Article in English | MEDLINE | ID: mdl-30133517

ABSTRACT

BACKGROUND AND OBJECTIVE: Complement activation as an early and important inflammatory process contributes to multiple organ dysfunction after trauma. We have recently shown that complement inhibition by decay-accelerating factor (DAF) protects brain from blast-overpressure (BOP)-induced damage. This study was conducted to determine the effect of DAF on acute lung injury induced by BOP exposure and to elucidate its possible mechanisms of action. METHODS: Anesthetized adult male Sprague-Daley rats were exposed to BOP (120 kPa) from a compressed air-driven shock tube. Rats were randomly assigned to three experimental groups: 1) Control (no BOP and no DAF treatment), 2) BOP (120 kPa BOP exposure), and 3) BOP followed by treatment with rhDAF (500µg/kg, i.v) at 30 minutes after blast. After a recovery period of 3, 24, or 48 hours, animals were euthanized followed by the collection of blood and tissues at each time point. Samples were subjected to the assessment of cytokines and histopathology as well as for the interaction of high-mobility-group box 1 (HMGB1) protein, NF-κB, receptor for advanced glycation end products (RAGE), C3a, and C3aR. RESULTS: BOP exposure significantly increased in the production of systemic pro- and anti-inflammatory cytokines, and obvious pathological changes as characterized by pulmonary edema, inflammation, endothelial damage and hemorrhage in the lungs. These alterations were ameliorated by early administration of rhDAF. The rhDAF treatment not only significantly reduced the expression levels of HMGB1, RAGE, NF-κB, C3a, and C3aR, but also reversed the interaction of C3a-C3aR and nuclear translocation of HMGB1 in the lungs. CONCLUSIONS: Our findings indicate that early administration of DAF efficiently inhibits systemic and local inflammation, and mitigates blast-induced lung injury. The underlying mechanism might be attributed to its potential modulation of C3a-C3aR-HMGB1-transcriptional factor axis. Therefore, complement and/or HMGB1 may be potential therapeutic targets in amelioration of acute lung injury after blast injury.


Subject(s)
Acute Lung Injury/drug therapy , Blast Injuries/drug therapy , CD55 Antigens/administration & dosage , HMGB1 Protein/genetics , Inflammation/drug therapy , Acute Lung Injury/genetics , Acute Lung Injury/physiopathology , Animals , Blast Injuries/genetics , Blast Injuries/pathology , Complement Activation/drug effects , Complement C3a/antagonists & inhibitors , Disease Models, Animal , Humans , Inflammation/genetics , Inflammation/physiopathology , Lung/drug effects , Lung/metabolism , Lung/physiopathology , NF-kappa B/genetics , Pressure/adverse effects , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...